Вы находитесь на странице: 1из 10

European Journal of Medicinal Chemistry 108 (2016) 79e88

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Anti-herpetic and anti-dengue activity of abietane ferruginol


analogues synthesized from (þ)-dehydroabietylamine
Vicky C. Roa-Linares a, b, Yaneth M. Brand a, b, Lee S. Agudelo-Gomez a,
 nica Tangarife-Castan
Vero ~ o a, Liliana A. Betancur-Galvis a, b, Juan C. Gallego-Gomez b,
lez c , *
Miguel A. Gonza
a
Group of Investigative Dermatology, Institute of Medical Research, Medicine Faculty, University of Antioquia, Medellin, A.A1226, Antioquia, Colombia
b
Translational and Molecular Medicine Group, Institute of Medical Research, Medicine Faculty, University of Antioquia, Medellin, Colombia
c
Departamento de Química Orga nica, Universidad de Valencia, E-46100 Burjassot, Valencia, Spain

a r t i c l e i n f o a b s t r a c t

Article history: The abietane-type diterpenoid (þ)-ferruginol (1), a bioactive compound isolated from several plants, has
Received 11 August 2015 attracted much attention as consequence of its pharmacological properties, which includes antibacterial,
Received in revised form antifungal, antimicrobial, cardioprotective, anti-oxidative, anti-plasmodial, leishmanicidal, anti-
23 September 2015
ulcerogenic, anti-inflammatory and antitumor actions. In this study, we report on the antiviral evalua-
Accepted 5 November 2015
Available online 11 November 2015
tion of ferruginol (1) and several analogues synthesized from commercial (þ)-dehydroabietylamine.
Thus, the activity against Human Herpesvirus type 1, Human Herpesvirus type 2 and Dengue Virus type
2, was studied. Two ferruginol analogues showed high antiviral selectivity index and reduced viral
Keywords:
Antiviral
plaque-size in post-infection stages against both Herpes and Dengue viruses. A promising lead, com-
Herpes pound 8, was ten-fold more potent (EC50 ¼ 1.4 mM) than the control ribavirin against Dengue Virus type
Dengue 2. Our findings suggest that the 12-hydroxyabieta-8,11,13-triene skeleton, which is characteristic of the
Abietane diterpenoid ferruginol (1), is an interesting molecular scaffold for development of novel antivirals. In
Diterpene addition, the cytotoxic and antifungal activities of the synthesized ferruginol analogues have also been
Ferruginol investigated. ©20155 Elsevier Science. All rights reserved.
Dehydroabietylamine © 2015 Elsevier Masson SAS. All rights reserved.

1. Introduction However, mutations in the viral DNA polymerase can also cause
this resistance. In immunocompromised patients, the presence of
Human Herpesvirus (HHV) types 1 and 2 are enveloped DNA HHV species resistant to ACV, with a prevalence of 4e10%, has
viruses characterized for producing latent infection in sensory neu- complicated their clinical management [3]. Moreover, Dengue virus
rons and reactivation during periods of severe immunosuppression (DENV) is an enveloped ssRNA virus. To date, there have been
of the host. Many factors such as stress, fatigue, sexual relations with described four serotypes established in humans (DENV-1 to DENV-
people who have active lesions, excess exposure to heat or cold, fe- 4) and recently, a fifth Dengue subtype that follows the sylvatic
ver, laser treatments, local tissue trauma and nerve damage, can cycle has been identified [4]. Dengue is the most important
predispose to reactivation, especially in neonates, transplant and mosquito-borne disease worldwide, since it is distributed over one
immunocompromised patients which tend to present aggressive and hundred countries of the tropical belt and the tropics, both the Old
recurrent lesions [1]. Also, it has been reported that prolonged and New World [5]. Disease manifestations cover a wide spectrum
therapy with Acyclovir (ACV), and its analogues, has induced the ranging from dengue with or without warning signs to severe
emergence of drug-resistant virus strains. Human Herpesvirus pre- dengue in which plasma leakage, haemorrhage and organ impair-
dominantly develops resistance, almost 95%, as a result of mutations ment can lead to death. In addition, unusual manifestations such as
in the genes that encode the viral thymidine kinase (TK) [2]. cardiomyopathy, liver failure and neurological disorders have been
reported [6]. The major problem of this pathology is that there are
no drugs approved for human treatment or vaccine available for
prophylaxis. Also, antiviral drugs addressed to specific genes or
* Corresponding author.
proteins of the virus are not a good alternative due to the high
E-mail address: Miguel.A.Gonzalez@uv.es (M.A. Gonz
alez).

http://dx.doi.org/10.1016/j.ejmech.2015.11.009
0223-5234/© 2015 Elsevier Masson SAS. All rights reserved.
80 V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88

mutation rate of RNA viruses, often because viral mutants are 6 gave 18-oxoferruginol (9) in moderate yield (50%), which was
selected for drug resistance [7]. During the last years in Colombia, converted into ferruginol (1) by Wolff-Kishner reduction (90%
several reports had shown the high rates of evolution of DENV, yield). And finally, the treatment of 6 with tetrachlorophthalic an-
possibly related with the new Asiatic-American genotypes found in hydride (TCPA) afforded phenol 10 in 75% yield, which was acety-
Colombia and Brazil [8]. lated with acetic anhydride in pyridine to give acetate 11 in
The aromatic abietane-type diterpenoids comprise a large quantitative yield. Acetate 11 was oxidized at C-7 with excess of t-
number of natural metabolites of plant origin which possess a wide BuOOH as oxidant and CrO3/pyridine mixture as a catalyst in DCM,
variety of biological effects [9]. These abietanes have been the the yield of ketone 12 was 66%. Subsequently, the reaction of 12
target of several synthetic campaigns towards both the natural with p-tosylhydrazide yielded the corresponding p-tosylhydrazone
products and synthetic derivatives with interesting pharmacolog- 13 (77% yield). The fullerene-terpenoid hybrid 14 was obtained by
ical properties [10]. Among these known bioactive compounds, the treatment with NaOMe of 13 in anhydrous pyridine for 20 min
(þ)-ferruginol (1), some derivatives of (þ)-dehydroabietic acid (2) at room temperature followed by addition of a solution of C60 in
and (þ)-dehydroabietylamine (3) as well as (þ)-jiadifenoic acid C chlorobenzene and heating at 70  C for 24 h. This gave compound
(4) (Fig. 1) have shown promising results, including antiviral 14 in ca. 30% yield. All the compounds showed spectroscopic data in
properties [10b,11]. agreement with the assigned structures and purity >95%.
The present study is a continuation of our research programs to
discover bioactive diterpenoids [12]. It includes the synthesis of a 2.2. Biological evaluation
focused library based on (þ)-ferruginol (1) from commercially
available (þ)-dehydroabietylamine (3), where a series of phthali- The synthesized compounds 1 and 5e14 (Scheme 1) were
mides could have potential antiviral activity [13]. A fullerene evaluated for antiviral activity against HHV-1, HHV-2 and DENV-2
diterpenoid hybrid was also envisaged as potential inhibitor [14]. (see Table 1). Two ferruginol analogues, compounds 8 and 9 (18-
Herein, we describe the antiviral activity of ferruginol (1) and some oxoferruginol), showed relevant activity.
analogues (5e14) (Scheme 1) synthesized from dehydroabietyl- In particular, the ferruginol analogue 8 showed moderate ac-
amine (3) against HHV-1 and HHV-2, and DENV-2. Our aim is to tivity against HHV-1 (Rf ¼ 1  102) at a concentration of 14.5 mM,
identify new antiviral drug candidates which act on cellular and/or high activity against HHV-2 (Rf ¼ 1  103) and reduction of cyto-
molecular targets instead on viral proteins. In this way, the con- pathic effect during DENV-2 infection, comparable to control of
cerns about the viral mutants resistant to the treatments can be untreated cells, at concentration of 29.0 mM. Compound 9 (18-
avoided. This approach has been recently called host-targeted oxoferruginol) also showed high activity against HHV-2
antiviral in an evolutionary study of resistance of Dengue viruses (Rf ¼ 1  103) at a concentration of 41.7 mM, however, its ability
under selective pressure of several antiviral agents [15]. to exert reduction of cytopathic effect in the presence of DENV-2
infection was lower than that of compound 8. Moreover, com-
pound 9 did not show activity against HHV-1.
2. Results and discussion
In 2007, Wen and co-workers reported the antiviral potential of
certain diterpenoid derivatives, including the abietane-type diter-
2.1. Chemistry
penoid ferruginol (1), during in vitro coronavirus infection [18].
However, at present, the antiviral activity of the abietane class of
The synthesis of (þ)-ferruginol (1) and analogues 5e14 from
compounds has not been much investigated, especially the study of
commercial (þ)-dehydroabietylamine (3) was performed as out-
derivatives and analogues; therefore, the activity against HHV-1,
lined in the Scheme 1. Compounds 1 and 5e9 were synthesized as
HHV-2 and DENV-2 reported in this study is to our knowledge,
reported in the literature using a FriedeleCrafts acylation and a
the first report of antiviral activity of ferruginol analogues. Also, as
BaeyereVilliger oxidation as key steps [16]. Then, compound 6 was
the compounds 8 and 9 are active against two different viruses,
used as starting material for the preparation of compounds 10 and
such as herpesvirus and dengue virus, our findings suggest that
11. Later, compound 11 was converted into compounds 12e14 as
these molecules probably have a broad-spectrum of antiviral ac-
described recently in the literature [17]. Thus, the synthesis starts
tivity against enveloped viruses with DNA and RNA genome.
with the introduction of the phthalimide group on (þ)-dehy-
Later on, to check the specific antiviral activity of these com-
droabietylamine (3), followed by FriedeleCrafts acylation and
pounds, we carried out the evaluation of other biological activities
oxidation under Baeyer-Villiger conditions to afford acetate 5 in
such as cytotoxic and antifungal, because is well known that fer-
72% overall yield. Hydrolysis of the acetate group in 5 gave phenol 8
ruginol (1) has promising bioactivities, such as antifungal, anti-
in high yield, while overall deprotection of 5 afforded the amino-
bacterial, miticidal antiplasmodial, antileishmanial, nematicidal,
phenol 6 in 75% yield. Compound 6 was the intermediate of three
and antitumor, among others [9]. In Table 2, it is shown the cyto-
separate approaches. Firstly, tosylation under standard conditions
toxic activity on Vero cells as well as on the tumor cells HeLa, Jurkat
gave compound 7 in 90% yield. Secondly, oxidative deamination of
and U937 of ferruginol 1 and analogues 5e14.
Compounds 1, 6, 7, 9, 10 and 12 produced a dose-dependent
inhibition on the growth of the three tumor cell lines: Jurkat,
U937 and HeLa, as well as the Vero cell line, with R2 (coefficient of
linear regression) > 0.8. Most of these compounds showed cyto-
toxic activity against at least one tumor cell line at concentrations
below 30 mM. However, only compound 10 against HeLa and Jurkat
cell lines, and compound 12 against the three tumor cell lines,
showed a great SI (SI  5), being compound 12 the most selective
agent against tumor cell lines respect the non-tumor cell line
(SI > 43.8, >26.0 and > 38.5, for HeLa, Jurkat and U937, respec-
tively). The compounds 5, 8, 11, 13 and 14 were not active against
the tumor cell lines at the tested concentrations and hence they
Fig. 1. Examples of bioactive aromatic abietane diterpenoids. were not evaluated on the Vero cell line.
V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88 81

Scheme 1. Synthesis of the tested compounds 1 and 5e14.

In addition, the antifungal activity of all compounds was studied Aspergillus fumigatus, Aspergillus flavus and Aspergillus terreus. Only
against two dermatophytes (Trichophyton rubrum and Trichophyton one ferruginol analogue, compound 9, showed anti-dermatophyte
mentagrophytes), and the filamentous fungi Fusarium oxysporum, activity against T. rubrum and T. mentagrophytes with geometric

Table 1
Reduction of viral titer, inhibition of cytopathic effect and antiviral activity against HHV-1, HHV-2 and DENV-2 of ferruginol analogues 8 and 9.

Vero cell linea

Compound HHV-1 HHV-2 DENV-2

Rfb Antiviral activity (mM)c Rfb Antiviral activity (mM)c CPE inhibitiond Antiviral activity (mM)c

8 102 14.5 103 29.0 þ 29.0


9 NA NA 103 41.7 þ 41.7
ACV 104 6.7 103 6.7 NT NT
RIBA NT NT NT NT þ 123.0

HHV-1: Human Herpesvirus type 1 (CDC Atlanta strain). HHV-2: Human Herpesvirus type 2 (VR-734-G strain). DENV-2: Dengue virus (New Guinea strain). NT: not tested. NA:
not active.
These values represent the mean of two independent experiments.
a
Vero Cercopithecus aethiops African green monkey kidney cell line ATCC CCL-81.
b
Reduction factor of the viral titer.
c
Maximal non-toxic concentration that showed viral reduction factor.
d
Cytopathic effect inhibition (þ).
82 V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88

Table 2
Cytotoxicity (IC50-mM) of ferruginol (1) and analogues 5e14 on HeLa, Jurkat, U937 and Vero cell lines.

Compound Cell linesc

HeLa Jurkat U937 Vero

IC50a (mM) SI b
IC50a (mM) SI b
IC50a (mM) SI b
IC50a (mM)

1 64.9 1.4 48.2 1.9 21.3 4.2 90.4


5 >50 NA >50 NA >50 NA NE
6 >80 NA 49.4 0.8 >80 NA 39.8
7 >50 NA 30.9 2.7 36.0 2.3 82.5
8 >50 NA >50 NA >50 NA NE
9 60.0 1.5 21.6 4.2 33.2 2.7 90.8
10 29.0 9.2 21.9 12.2 >40 NA 266.8
11 >40 NA >40 NA >40 NA NE
12 7.3 >43.8 12.3 >26.0 8.3 >38.5 >320
13 >30 NA >30 NA >30 NA NE
14 >30 NA >30 NA >30 NA NE
PXT 0.099 NA 0.0005 NA 0.006 NA 0.65
DOX 0.834 NA 0.012 NA 0.077 NA 1.93

PXT: Paclitaxel; DOX: Doxorubicine. NA: Not apply; NE: Not evaluated. These values represent the geometric mean of two independent experiments, each one made by
quadruplicate.
a
Inhibitory concentration 50 defined as concentration of compounds that induces 50% of growth inhibition at 48 h.
b
SI, selectivity index defined as VERO IC50 over either Jurkat, U937 or HeLa IC50.
c
Jurkat, human acute T cell leukemia ATCC TIB-152; U937, human promonocytic cell line ATCC CRL-1593.2; HeLa, human cervix epitheloid adenocarcinoma cells ATCC CRL-
1958; Vero, Cercopithecus aethiops African green monkey kidney cells ATCC CCL-81.

mean of minimal inhibitory concentration (GM-MIC) values of between the inhibitory concentration 50 (IC50) in Vero cells and
41.7 mM and 83.3 mM, respectively. None of the compounds showed EC50 of each virus (IC50/EC50) (Table 3).
activity against the filamentous fungus tested (data not shown). Compound 8 reduced the 50% of plaque forming units at a
Likewise, none of the compounds showed anti-Candida activity concentration of 19.2 mM against HHV-2 and showed a high SI equal
against three yeast strains (Candida albicans, Candida parapsilosis to 10, whereas compound 9 reduced the number of plaques in 50%
and Candida tropicalis) (data not shown). at concentrations of 19.6 mM and 16.6 mM, for HHV-1 and HHV-2,
Although there are several studies showing that ferruginol (1) respectively. As expected the controls, DEX-S and ACV, did show
and some related abietanes exhibit promising antifungal and antiviral activity in pre-treatment and post-treatment, respectively.
cytotoxic activity [19], our results reveals that compound 8 lacks of These results show that the compounds 8 and 9 have important
antifungal activity in the fungal models tested and cytotoxicity activity against HHV-2 strain at low concentrations, in comparison
against HeLa, Jurkat and U937 tumor cells at the evaluated con- to HHV-1. This is possibly due to a smaller number of receptors
centrations. Therefore, this compound could be a selective mole- available in the host cell to carry out the infection with HHV-2,
cule against enveloped viruses. making this virus more sensitive to the action of the compounds
[21].
2.3. Mechanism of antiviral activity of ferruginol analogues 8 and 9 Differences in the activity of these compounds were found
against HHV-1 strain. In the end-point titration technique (EPTT)
Previous studies suggest that the mechanism by which ferru- assay, compound 8 showed moderate activity (Table 1, Rf ¼ 1  102)
ginol (1) exerts antitumor activity is related to the enhanced and in the plaque forming unit (PFU) assay this compound did not
expression of BAX proapoptotic protein, inhibition of signaling show a dose-dependent effect. However, it showed a significant
pathways involved in cell survival and proliferation (Ras /PI3K and reduction in viral plaque size (Table 3, highlighted with an asterisk),
Jak/STAT) and reduction of expression of cyclin-dependent kinases related among others, with a low rate of viral replication and
[20]. However, the cellular and molecular basis by which these reduction of cytopathic effect, confirming the antiviral effect of this
compounds exhibit antiviral activity has not been currently compound.
described. On the other hand, compound 9 did not show reduction of viral
In order to define the stage of viral replication cycle in which the titer during screening (EPTT) against HHV-1, but inhibits the
ferruginol analogues 8 and 9 were active, we performed two number of plaque forming units during the PFU assay. This differ-
different conditions of treatment. Firstly, we tested the effect of ence can be explained due the amount of virus used in each assay.
compounds on early stages of viral replication cycle, such as cell- In the EPTT assay a high amount of virus (10 TCID50) was used and
free viral particle, adhesion and viral entry by treatment before the concentration of compound that showed antiviral activity could
infection (pre-infection treatment). And secondly, we tested the not be detected. In the PFU assay, 100 PFU/well was used and the
antiviral activity on several steps of viral cycle that includes traffic, EC50 value was determined, nevertheless, this compound did not
replication, assembly, egress of new viral particles and cell-to-cell show a high SI value (4.9), demonstrating closeness between the
spread by treatment after infection (post-infection treatment). cytotoxic concentration and the antiviral concentration.
As can be seen in Table 3, compounds 8 and 9 were effective Likewise, the anti-dengue activity of these ferruginol analogues
during post-infection treatment and were not active during pre- was evaluated 8 days post-infection (d.p.i), being both compounds
infection treatment. 8 and 9 active at EC50 concentrations of 1.4 mM (SI ¼ 57.7) and
With both pre- and post-infection treatments, a plaque reduc- 5.0 mM (SI ¼ 10.4), respectively.
tion assay was performed and the concentration of compounds 8 The antiviral selectivity index is defined as the relative effec-
and 9 that reduced the number of viral plaques in 50% (EC50) was tiveness of a product to inhibit viral replication compared to the
interpolated from the doseeresponse curves. Furthermore, the capacity for inducing cell death. This index results useful to make
antiviral selectivity index (SI) was calculated through the relation bioactivity comparisons between the compounds and helps in
V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88 83

Table 3
Antiviral activity (EC50-mM) on pre and post-infection treatment of ferruginol analogues 8 and 9.

Compound IC50 IC50 EC50 (mM) (SI) EC50 (mM) (SI)


(mM) (mM) Pre-infection treatment Post-infection treatment
72 h 8 days
HHV-1 HHV-2 DENV-2 HHV-1 HHV-2 DENV-2

8 190.7 80.8 NI NI NI NI* 19.2 (10.0) 1.4 (57.7)*


9 97.8 52.2 NI NI NI 19.6 (4.9) 16.6 (5.9)* 5.0 (10.4)
DEX-S >10 NT 0.015 0.012 NT NT NT NT
ACV >1700 NT NT NT NT 2.2 0.27 NT
RIBA NT NT NT NT NT NT NT 13.5

EC50, 50% antiviral effective concentration; IC50, 50% inhibitory concentration of cell proliferation (mM); SI, Antiviral selectivity index values (IC50/EC50) are presented between
parenthesis; DEX-S, dextran sulfate; ACV, acyclovir; RIBA, ribavirin; *Reduction on viral plaque-size. NI, no inhibitory activity; NT, not tested. These values represent the mean
of three independent experiments.

designing more potent compounds. In recent reports, SI values verified 24 h post-treatment using a fluorescent plaque assay in the
higher than 10 (SI > 10) are considered indicative of a potential presence of HHV-1 neutralizing antibodies to guarantee cell-to-cell
therapeutic agent that would merit further biopharmaceutical and transmission via intercellular junctions between infected and un-
pre-clinical studies [22]. Therefore, compound 8 is promissory to infected cells [24]. Meanwhile, Nyberg and Ekblad and co-workers
continue this type of study. demonstrated that the low molecular weight heparin sulfate-
Additionally to the dose-dependent effect previously described mimic, PI-88, which is a mixture of highly sulfated mannose con-
during post-infection treatment, we found that compounds 8 and 9 taining oligosaccharides, reduced the area of viral plaques with IC50
induced a reduction on viral plaque size. To determine the average values of 2 mg/mL and 0.7 mg/mL for HHV-1 and HHV-2, respectively
of reduction of viral plaque size during compound-treatment [25]. Nevertheless, to date it has not been reported a similar effect
relative to mock-treated control, we performed an image analysis on viral plaque size of HHV and other viruses from abietane diter-
using Image-Pro Plus 6.0 software (Fig. 2). The area of plaques was penoids, among these, ferruginol (1) and their derivatives.
measured in millimeters and statistically significant differences (P Several molecular and cellular components involved in
value < 0.001) were found in all cases. Compound 8 showed a Herpesvirus cell-to-cell spread have been previously reported. For
dramatic reduction on viral plaque size (93.7%) at a concentration example, viral glycoproteins such as E and M play a relevant role in
of 29.0 mM against HHV-1 (CDC-Atlanta acyclovir-sensitive strain) normal viral plaque phenotype of HHV-1, because the plaque size of
(Fig. 2A). Respect to HHV-2, compound 9 reduced the size of viral the double-deletion mutant (DgEgM) reduced over 5-fold less than
plaques (68.2%) at a concentration of 20.8 mM (Fig. 2B). wild type strain [26]. In polarized epithelial cells, Johnson and co-
Furthermore, during treatment with compound 8 at a concen- workers demonstrated by electron microscopy, that the gE/gI gly-
tration of 29.0 mM (Fig. 2C) on cell infection by HHV-1 acyclovir- coproteins complex is necessary to accumulate virions at cell
resistant strain 29R (thymidine kinase mutant), we found a similar junctions, since mutants that not express gE glycoprotein leads to
effect to that showed against the acyclovir-sensitive strain, particles accumulation more frequently on apical surfaces. Thus,
reducing the viral plaque size in 87.1%. This finding may suggest they concluded that the gE/gI complex is required to sort selectively
that the phosphorylation by viral thymidine kinase it is not HHV-1 particles to lateral surfaces and specifically to cell junctions
required for the effect exerted by this compound, thus, is possible [27]. On the other hand, structures containing actin and tubulin
that anti-herpetic mechanism of action of ferruginol analogue 8 during Herpesviruses infection have the ability to project virions
differs to acyclovir and other nucleoside analogues. towards adjacent cells. Additionally, both the membrane compo-
Likewise, reduction on viral plaque-size (77.9%) was observed sition and cytoskeletal structures of egress sites are modified dur-
when cells infected with DENV-2 were treated with compound 8 at ing infection in non-polarized cells [28]. According to the findings
concentration of 3.6 mM (Fig. 2D). The reduction percentages re- of Dixit and co-workers, HHV-1 also promotes cytoskeletal rear-
ported for all viruses indicate the reduction of viral plaque size rangements that facilitate viral spread in vitro, including dramatic
during compound-treatment relative to mock-treated control increases of filopodia in cultured neurons [29]. Multiple and
(100%). Representations of plaque morphology of HHV-1(CDC convergent ways by which several viruses such as Vaccinia, Hepa-
Atlanta), HHV-1 (29R) and DENV-2 after the treatment with com- titis B and C viruses, Human Immunodeficiency virus, Human T-
pound 8 and mock-treated control, are shown in Figures 2E, F and lymphotropic virus, Murine leukemia virus and HHV cause sub-
2H, respectively. Fig. 2G shows the effect after treatment with version of cytoskeleton for different objectives in viral cycle are
compound 9 against HHV-2 and mock-treated control. summarized by Taylor and co-workers [30]. The actin cytoskeleton
The reduction on viral plaque size of HHV strains and DENV-2 in is reorganized by these viruses affecting every stage of the viral life
the presence of compounds 8 and 9 suggests that these ferruginol cycle, from entry through assembly to egress. Using immuno-
analogues may prevent the first-round of viral replication, release chemical assays, drug inhibition assays and protein interaction
and/or cell-to-cell spread, being this last stage an important factor profiling methods, Wang and co-workers identified the ways in
in infectivity, virulence, establishment of latency and immune which DENV-2 interacts with actin cytoskeleton. This study showed
response evasion during Herpesvirus infections [23]. Therefore, that dynamic treadmilling of actin is necessary for Dengue virus
inhibition of viral intercellular diffusion is an attractive target in the entry, production and release, indicating a direct effect of viral E
search for new antiviral drugs, mainly to treat the latent infections protein on the structural modifications of actin cytoskeleton [31].
and drug-resistant strains. Considering that cytoskeletal components such as microtubules
In recent years, several compounds have shown effects on virus and actin microfilaments are required for both Herpesvirus and
cell-to-cell spreading during Herpesvirus infection. For example, Flavivirus replication cycle (mainly involved in the entry, traffic,
Lin and co-workers reported that chebulagic acid and punicalagin, release and cell-to-cell spread), is possible that ferruginol ana-
two hydrolyzable tannins isolated from of Terminalia chebula logues 8 and 9 cause disruption of these cellular pathways which
(Combretaceae), inhibit the HHV-1 cell-to-cell spread. This fact was may be involved in viral spread. Nonetheless, other experiments
Fig. 2. Effect of compounds 8 and 9 on HHV-1, HHV-2 and DENV-2 plaque-size during Vero cell infection. Effect on viral plaque-size of compounds 8 and 9 in presence of 100 PFU/
mL of HHV-1 (2A), HHV-2 (2B), HHV-1 29R strain (2C) and DENV-2 (2D). Results were expressed as an average of plaque size area (mm) of 80 viral plaques developed in compound-
treated cells relative to mock-treated controls. Statistically significant differences were found in all cases with p values < 0.001 (***). Parallel, the image analysis using Image-Pro
Plus 6.0 software represents the reduction of viral plaque size during treatment with compounds 8 and 9 of HHV-1 (2E), HHV-2 (2F), HHV-1 29R strain (2G) and DENV-2 (2H). Two
separate experiments were carried out for each compound.
V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88 85

are required to confirm this hypothesis. procedures [16]. Compound 6 was used as starting material for the
Although it is very risky assure that some viral or cellular preparation of compounds 10 and 11. Later, compound 11 was
component are involved in our findings, the study of ferruginol converted into compounds 12e14 as described recently in the
analogues will be an important tool from medicinal chemistry for literature [17]. All compounds prepared in this work exhibit spec-
dissecting these complicated cellular and molecular processes of troscopic data in agreement with the proposed structures. Purity of
the viral infections. In the future, this research will be routed to all final compounds was 95% or higher.
understanding of some cellular and molecular mechanisms where,
two very different viruses (Herpesvirus and Dengue), are 4.1.2.1. 12-Hydroxy-N,N- (tetrachlorophthaloyl)
converging in a conserved evolutionary pathway during cell dehydroabietylamine (10)
infection, affecting cytoskeleton elements, endomembrane system A mixture of the tetrachlorophthalic anhydride (640 mg,
and signaling pathways involved in viral egress from the host-cells 2.14 mmol) and 12-hydroxydehydroabietylamine 6 [16a] (647 mg,
and spreading infection toward neighbor cells. 2.14 mmol) in acetic acid (7.5 mL) was heated at reflux for 2 h, then
cooled to rt and 50 mL of water was added. The resulting solid was
3. Conclusions filtered off, washed with water, dried under vacuum and purified by
column chromatography eluting with toluene to give compound 10
In summary, we have discovered important antiviral properties (913 mg, 75%) as a yellow solid: [a]20 1
D 10.0 (c 1.0, CHCl3); H NMR
in the molecular scaffold 12-hydroxyabieta-8,11,13-triene func- (300 MHz) d 6.87 (1H, s), 6.60 (1H, s), 3.67 (1H, d, J ¼ 15.0), 3.54 (1H,
tionalized at C-18, which is reminiscent of the abietane diterpenoid d, J ¼ 15.0), 3.10 (1H, m), 2.92 (2H, m), 2.15 (2H, m), 1.24 (3H, d,
ferruginol (1). Two ferruginol analogues, compounds 8 and 9, J ¼ 6.0), 1.22 (3H, d, J ¼ 6.0), 1.22 (3H, s), 1.04 (3H, s); 13C NMR
synthesized from readily available (þ)-dehydroabietylamine (3) (75 MHz) dC 2  164.5 (s), 150.6 (s), 148.1 (s), 2  140.1 (s), 131.6 (s),
were found to consistently inhibit Human Herpesvirus (type 1 and 2  129.5 (s), 2  127.4 (s), 127.1 (s), 126.8 (d), 110.5 (d), 49.8 (t), 45.2
2) and Dengue virus type 2 at low micromolar concentrations. (d), 39.5 (s), 38.0 (t), 37.5 (s), 37.1 (t), 29.3 (t), 26.7 (d), 25.7 (q), 22.7
These compounds showed antiviral activity specifically in post- (q), 22.5 (q), 19.5 (t), 19.0 (q), 18.4 (t). HRMS (ESI) m/z 568.0986
infective stages and significantly reduced the viral plaque-size [Mþ1]þ, calcd for C28H30Cl4NO3: 568.0980.
during infection. The presence of a phthalimide moiety in the
molecule, compound 8, resulted in the highest antiviral selectivity 4.1.2.2. 12-Acetoxy-N,N-(tetrachlorophthaloyl)
index against the viruses tested and a ten-fold more potency dehydroabietylamine (11)
(EC50 ¼ 1.4 mM) than the control ribavirin against Dengue Virus A solution of phenol 10 (656 mg, 1.15 mmol) in a 1:1 mixture
type 2. (14 mL) of acetic anhydride and pyridine was stirred at rt for one
With regard to the cytotoxic properties of the tested com- day. Then, 150 mL of water were added and the mixture was
pounds, it should be noted that the chlorinated-phthalimide extracted with ethyl acetate (3  25 mL). The combined organic
analogue 12 exhibited moderate cytotoxicity of broad spectrum, extracts were washed with 6% NaHCO3 (2  50 mL), water and
without toxicity towards the non-tumor cell line (Vero). Addi- brine. The resulting organic extract was dried over sodium sulfate,
tionally, the tested compounds did not show relevant antifungal filtered and concentrated. The residue was chromatographed on
activity. silica eluting with chloroform to afford acetate 11 (704 mg, 100%) as
Based on the above, the results here reported are relevant for a white solid with identical spectroscopic data to the reported in
further research of possible cellular and molecular mechanisms the literature [17].
involved in the effect of reduction on viral plaque size. Also, these
findings are important to promote the realization of biopharma- 4.3. Biology
ceutical testing and finding new highly selective antiviral drugs
mainly directed towards cellular targets. 4.3.1. Reagents and compounds
Dulbecco's Modified Eagle's Medium (DMEM) and 3-(4,5-
4. Experimental dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT)
were obtained from SigmaeAldrich Chemical Co. (St. Louis, MO,
4.1. Chemistry USA). Fetal bovine serum (FBS) and penicillin/streptomycin were
purchased from Invitrogen Life Technologies (Carlsbad, CA, USA).
4.1.1. General procedures Acyclovir was obtained from Biogen Laboratory. Ribavirin was ob-
Optical rotations were measured using a 5 cm cell in a Schmidt- tained from Calbiochem (La Jolla, CA, USA). Paclitaxel, Doxor-
Haensch Polartronic-D polarimeter. NMR spectra were recorded on ubicine, Amphotericine B and Itraconazole were purchased from
a 300 MHz spectrometer. All spectra were recorded in CDCl3 as SigmaeAldrich Chemical Co. Terbinafine was obtained from
solvent unless otherwise stated. Complete assignments of 13C NMR Recalcine Laboratories (Santiago de Chile, Chile). Dimethyl sulf-
multiplicities were made on the basis of DEPT experiments. J values oxide (DMSO) was purchased from Merck KGaA (Darmstadt,
are given in Hz. MS data were acquired on a QTOF spectrometer. Germany).
Reactions were monitored by TLC using Merck silica gel 60 F-254 in Stock solutions of compounds were prepared in DMSO and
0.25 mm-thick plates. Compounds on TLC plates were detected frozen at 70  C. The concentration of DMSO in biological assays
under UV light at 254 nm and visualized by immersion in a 10% was of 0.05%. Cell controls with DMSO at 0.05% were used.
sulfuric acid solution and heating with a heat gun. Purifications
were performed by flash chromatography on Merck silica gel 4.3.2. Cell culture and viruses
(230e400 mesh). Commercial reagent grade solvents and chem- Human Herpesvirus type 1 (HHV-1 CDC Atlanta acyclovir-
icals were used as purchased unless otherwise noted. Combined sensitive strain) was obtained from the Center for Disease Control
organic extracts were washed with brine, dried over anhydrous (Atlanta, GA, USA); HHV-1 29R (acyclovir-resistant strain) was
NaSO4, filtered, and concentrated under reduced pressure. donated by Prof. Claudia Oliveira (University of Santacatarina-
Brazil) and Human Herpesvirus type 2 (HHV-2 VR-734-G
4.1.2. Materials acyclovir-sensitive strain) was obtained from the Center for Dis-
Compounds 1 and 5e9 have been prepared according to known ease Control. All of them were amplified in Vero cells (African green
86 V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88

monkey kidney-Cercopithecus aethiops, ATCC: CCL 81 line). Virus monolayer. The cell monolayers were stained with a solution of
stocks were titrated in Vero cells by plaque assay and expressed as 3.5% formaldehyde with 0.2% crystal violet and CPE was observed
plaque forming units (PFU/mL). Dengue virus type 2 (DENV-2 New under inverted microscope. Controls were included untreated cells,
Guinea strain) was donated by Maria Elena Pen ~ aranda and Eva cells treated with compounds and cells infected with DENV-2. As
Harris (Sustainable Sciences Institute and the University of Cali- positive control we used Ribavirin. Compounds were considered
fornia at Berkeley) was amplified in C6/36HT of Aedes albopictus active against DENV-2 if has the ability to reduce or avoid the
cells, from ATCC, and titrated in Vero cells following our laboratory cytopathic effect during viral infection. This assay was conducted
conditions [32]. by quadruplicate and repeated twice for each compound.
Cells were maintained in Dulbecco's Modified Eagle Medium
(DMEM) supplemented with 5% of inactivated FBS to Vero cells and 4.4.3. Assays for pre- and post-infective stages of HHV-1, HHV-2
10% of inactivated FBS to C6/36HT cells, 100 units/mL of penicillin, and DENV-2
100 mg/mL of streptomycin, 100 mg/mL of L-glutamine, 0.14% Subsequently, the potential antiviral of active compounds was
NaHCO3, and 1% of each non-essential amino acids and minimum evaluated by the plaque reduction assay as previously described
essential medium vitamin solution (choline chloride, D-calcium [35]. Vero cell monolayers grown in 24-well plates (2.5  104 cells/
pantothenate, folic acid, nicotinamide, pyridoxal hydrochloride, well) were infected with 100 PFU per well of each virus. Treatments
riboflavin, thiamine hydrochloride and i-inositol). Vero cells were were performed by adding compounds either simultaneously with
incubated at 37  C in humidified 5% CO2 atmosphere and C6/36 HT virus (pre-infection treatment) or after viral infection (post-infection
at 34  C in humidified 5% CO2 atmosphere. treatment). For pre-infection treatment, a virus/compound mixture
To determine the cytotoxic activity, cell lines of human cervix was added to cell monolayers and was incubated for 1 h at 37  C (5%
epitheloid adenocarcinoma cells (HeLa, ATCC CCL-2), acute T cell CO2). After incubation, washing was performed with PBS (pH ¼ 7.0)
leukemia (Jurkat, ATCC TIB-152) and human promonocytic cell line and added CMC 1%, 0.75% and 2% for HHV-1, HHV-2 and DENV-2,
(U937, ATCC CRL-1593.2) were used, as well as, the non-tumor cell respectively. In post-infection treatment, virus was added on cell
line Vero. Vero and HeLa cells were grown in DMEM supplemented. monolayer and incubated for 1 h at 37  C (5% CO2). After, washing
Jurkat and U937 cells were maintained in RPMI-1640 medium was performed with PBS (pH ¼ 7.0) and compounds previously
(supplemented with 10% FBS), 100 units/mL of penicillin, 100 mg/mL prepared in CMC 1%, 0.75% and 2% for HHV-1, HHV-2 and DENV-2
of streptomycin, and 100 mg/mL of neomycin and maintained at respectively, were added. In both treatments, plates allowed to
37  C in humidified 5% CO2 atmosphere. incubate for 72 h (HHV-1 and HHV-2) and 8 days (DENV-2). Cells
were then fixed and stained with a solution of 3.5% formaldehyde
4.4. Antiviral activity with 0.2% crystal violet and viral plaques were counted. Dextran
sulfate was employed as positive control in pre-infection assays.
4.4.1. Screening against HHV-1 and HHV-2 Ayclovir (ACV) and Ribavirin (RIBA) were used as positive controls
The anti-herpetic activity of ferruginol analogues on Vero cells in post-infection stages to HHV strains and DENV-2, respectively.
against 10 TCID50 of HHV-1 and HHV-2, were carried out using the The fifty effective concentration (EC50) was defined as the con-
end-point titration technique (EPTT). Vero cells were harvested in centration that reduces the 50% of plaque forming units. EC50 for
96-well plates at a density of 2.0  104 cells/well at 37  C in hu- each compound were obtained from doseeeffect curves for linear
midified 5% CO2 atmosphere until constituted 80% of the cell regression methods using the statistical GraphPad Prisma 5.0 and
monolayer. Viral suspension/compound mixture were performed EC50 values are expressed as the mean of at least four dilutions by
in DMEM with 2% FBS supplemented containing carboxymethyl- quadruplicate. To define which compounds were more selective to
cellulose (CMC) to 1% and 0.5% for HHV-1 and HHV-2, respectively infected cells than to non-infected cells, the antiviral selectivity
and incubated for 15 min at room temperature. Compounds were index (SI) was calculated and defined as the ratio between the
evaluated at concentrations of 6.25e50 mg/mL. After 48 h (HHV-1) inhibitory concentration 50 (IC50) in Vero cells and the EC50 for each
and 72 h (HHV-2) of incubation at 37  C in a humidified 5% CO2 virus on Vero cells.
atmosphere, the cell monolayers were stained with a solution of
3.5% formaldehyde with 0.2% crystal violet and the plaques were 4.4.4. Plaque size-reduction assay
counted. Two independent experiments by quadruplicate were For this, a PFU assay was carried out on Vero cells
carried out for each viral serotype and each compound. Controls (2.5  104 cells/well) and compounds previously prepared in CMC,
were included untreated cells, cells treated with compounds and were added 2 h after infection with 100 PFU/well of virus [25b].
cells infected with HHV-1 or HHV-2. Positive control included in Compound 8 was added at concentrations from 1.56 mg/mL to
this assay was Acyclovir. According to the parameters established 12.5 mg/mL and compound 9 at concentrations from 0.78 mg/mL to
by Vlietinck et al. [33] the relevant or moderate antiviral activity of 6.25 mg/mL, against both HHV-1 (CDC-Atlanta and 29R) and HHV-2.
a purified natural product, is one whose reduction factor (Rf) of viral To determine the effect against DENV-2, compounds 8 and 9 were
titer is respectively of  1  103 or 1  102. This parameter was used added in a concentration range of 0.05 mg/mL to 1.56 mg/mL. Finally,
in our study. the plates were incubated for 72 h (HHV strains) and 8 days (DENV-
2), fixed and stained with a solution of 3.5% formaldehyde/0.2%
4.4.2. Screening against DENV-2 crystal violet. The images of 20 plaques for each compound con-
To determine the anti-DENV activity we carried out the protocol centration and each well were captured using a Nikon digital
previously described by Tang et al. [34] with some modifications. camera view DS-L1 attached to a Nikon inverted microscope
Briefly, Vero cells were harvested in 96-well plates at a density of (Tokyo, Japan) [25a]. The area of each plaque in millimeters was
2.0  104 cells/well at 37  C in humidified 5% CO2 atmosphere. After determined using the Image-Pro Plus 6.0 software®. Two inde-
24 h, medium was removed and two-fold serial dilutions of DENV-2 pendent experiments were performed by duplicate.
at 10TCID50 and non-cytotoxic concentrations of compounds were
incubated for 10 min at room temperature. Then, 50 mL of each 4.4.5. Cytotoxic activity
dilution (compound/DENV-2) was added and the plates were Cell growth inhibition and/or cytotoxicity on Vero, HeLa, Jurkat
incubated at 37  C for 3 days, time at which the cytopathic effect and U937 cells were measured using the technique MTT. The MTT
(CPE) in control (infected cells) was observed in 100% of cell 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88 87

(Sigma, New Jersey, USA) assay, according to the protocol previ- Financiamiento para la Ciencia, la Tecnología y la Innovacio n,
ously described [12c] with few modifications, was used. Briefly,  de Caldas) is gratefully acknowledged. V. C. R.-L., L. S.
Francisco Jose
Vero and HeLa cells were plated at 2.0  104 cells per well in a 96- A.-G. and V. T.-C. thank COLCIENCIAS Programa Jo  venes Inves-
well flat-bottomed plates and incubated for 24 h at 37  C. Moreover, tigadores for their fellowships.
Jurkat and U937 cells were plated at 3.0  104 cells per well in a 96-
well round-bottomed plate. Then each diluted compound was References
added to the appropriate wells and the plates were incubated for
further 48 h at 37  C in a humidified incubator with 5% CO2. Finally, [1] D.D. Richman, R.J. Whitley, F.G. Hayden, Clinical Virology, third ed., ASM Press,
Washington, DC, 2009.
spectrophotometric reading was carried out at 570 nm to deter- [2] J. Piret, G. Boivin, Antiviral drug resistance in herpesviruses other than cyto-
mine the inhibitory concentration 50 (IC50), defined as the con- megalovirus, Rev. Med. Virol. 24 (2014) 186e218.
centration for each compound that inhibits 50% of cell growth. The [3] J. Piret, G. Boivin, Resistance of herpes simplex viruses to nucleoside ana-
logues: mechanisms, prevalence, and management, Antimicrob. Agents Che-
IC50 values for each compound were obtained by linear regression
mother. 55 (2011) 459e472.
analysis of the doseeresponse curves generated from the absor- [4] M.S. Mustafa, V. Rasotgi, S. Jain, V. Gupta, Discovery of fifth serotype of dengue
bance data with the statistical GraphPad Prisma 5.0. IC50 values virus (DENV-5): a new public health dilemma in dengue control, Med. J.
were expressed as the geometric mean of two independent ex- Armed Forces India 71 (2015) 67e70.
[5] K.A. Hanley, S.C. Weaver, Frontiers in Dengue Virus Research, Caister Aca-
periments done in quadruplicate. To define which compounds were demic Press, Norfolk, 2010.
more selective to cancerous cells than to non-cancerous cells, it was [6] TDR/WHO, Dengue: guidelines for diagnosis, treatment, Prevention and
calculated selectivity index (SI) defined as Vero IC50 over HeLa, Control, World Health Organization, Geneva, 2009.
[7] M. De Wispelaere, A.J. LaCroix, P.L. Yang, The small molecules AZD0530 and
Jurkat or U937 IC50. A compound with an IC50 < 30 mM was dasatinib inhibit dengue virus RNA replication via fyn kinase, J. Virol. 87
considered active and with a SI  5 was considered selective. (2013) 7367e7381.
Paclitaxel and Doxorubicine were used as positive controls. [8] (a) J.A. Usme-Ciro, J.A. Mendez, A. Tenorio, G.J. Rey, C. Domingo, J.C. Gallego-
Gomez, Simultaneous circulation of genotypes I and III of dengue virus 3 in
Colombia, Virol. J. 5 (2008) 101;
4.4.6. Antifungal activity (b) J.A. Mendez, J.A. Usme-Ciro, C. Domingo, G.J. Rey, J.A. Sanchez, A. Tenorio,
The in vitro antifungal activity of compounds were evaluated J.C. Gallego-Gomez, Phylogenetic history demonstrates two different lineages
of dengue type 1 virus in Colombia, Virol. J. 7 (2010) 226.
following the Clinical and Laboratory Standards Institute M38-A [9] M.A. Gonza lez, Aromatic abietane diterpenoids: their biological activity and
protocol for filamentous fungi [36] with a few modifications and synthesis, Nat. Prod. Rep. 32 (2015) 684e704.
the standard method proposed by the Antifungal Susceptibility [10] (a) M.A. Gonza lez, Aromatic abietane diterpenoids: total syntheses and syn-
thetic studies, Tetrahedron 71 (2015) 1883e1908;
Testing Subcommittee of the European Committee on Antibiotic
(b) M.A. Gonz alez, Synthetic derivatives of aromatic abietane diterpenoids
Susceptibility Testing (AFST-EUCAST) for yeasts [37]. The filamen- and their biological activities, Eur. J. Med. Chem. 87 (2014) 834e842.
tous fungi F. oxysporum (ATCC 48112), A. fumigatus (ATCC 204305), [11] G.-J. Zhang, Y.-H. Li, J.-D. Jiang, S.-S. Yu, J. Qu, S.-G. Ma, Y.-B. Liu, D.-Q. Yu, Anti-
A. flavus (ATCC 204304), A. terreus (CDC 317) and dermatophytes, T. Coxsackie virus B diterpenes from the roots of Illicium jiadifengpi, Tetrahe-
dron 69 (2013) 1017e1023.
rubrum (ATCC 28188) and T. mentagrophytes (ATCC 24198) were [12] (a) L. Betancur-Galvis, C. Zuluaga, M. Arno , M.A. Gonz alez, R.J. Zaragoz a,
used to evaluate antifungal activity at inoculum size of Structure-activity relationship of in vitro antiviral and cytotoxic activity of
0.2e2.5  105 CFU/mL. The yeasts C. albicans (ATCC 1023), C. par- semisynthetic analogues of scopadulane diterpenes, J. Nat. Prod. 64 (2001)
1318e1321;
apsilosis (ATCC 22019) and C. tropicalis (ATCC 200956) were used to (b) M.A. Gonza lez, Spongiane diterpenoids, Curr. Bioact. Comp. 3 (2007) 1e36;
evaluate antifungal activity at inoculum size of 1e5  105 CFU/mL. (c) B. Zapata, M. Rojas, L. Betancur-Galvis, A.C. Mesa-Arango, D. Pe rez-Guaita,
The dilutions of compounds were dispensed into 96-well flat- M.A. Gonza lez, Cytotoxic, immunomodulatory, antimycotic, and antiviral ac-
tivities of semisynthetic 14-hydroxyabietane derivatives and triptoquinone C-
bottom microdilution plates in duplicate at final concentrations 4 epimers, Med. Chem. Comm. 4 (2013) 1239e1246.
between 50 mg/mL and 3.125 mg/mL. The compounds were [13] J. Balzarini, E. De Clercq, B. Kaminska, A. Orzeszko, Synthesis and antiviral
considered active when they exhibited MIC values  50 mg/mL. For activity of some 50 -N-phthaloyl-30 -azido-20 ,30 -dideoxythymidine analogues,
Antivir. Chem. Chemother. 14 (2003) 139e144.
the CLSI M38-A method, the MIC were defined as the lowest dilu-
[14] R. Bakry, R.M. Vallant, M. Najam-ul-Haq, M. Rainer, Z. Szabo, C.W. Huck,
tion that resulted in an 80% of inhibition of visible growth after G.K. Bonn, Medicinal applications of fullerenes, Int. J. Nanomed. 2 (2007)
incubation at 28  C to 48 h to F. oxysporum and 6 days to derma- 639e649.
[15] E. Plummer, M.D. Buck, M. Sanchez, J.A. Greenbaum, J. Turner, R. Grewal,
tophytes. Amphotericine B was evaluated, at a range of 0.031e16
B. Klose, A. Sampath, K.L. Warfield, B. Peters, U. Ramstedt, S. Shresta, Dengue
mg/mL, with the strains A. fumigatus (ATCC 204305) and A. flavus virus evolution under a host-targeted antiviral, J. Virol. 89 (2015) 5592e5601.
(ATCC 204304) and Terbinafine was used as positive control at a [16] (a) M.A. Gonz alez, D. Perez-Guaita, Short syntheses of (þ)-ferruginol from
range of 0.0078e4 mg/mL on both dermatophytes. For the AFST- (þ)-dehydroabietylamine, Tetrahedron 68 (2012) 9612e9615;
(b) I.M. Malkowsky, M. Nieger, O. Kataeva, S.R. Waldvogel, Synthesis and
EUCAST method, the MIC was determined after 24 h of incuba- properties of optically pure phenols derived from (þ)-dehydroabietylamine,
tion at 35  C and defined as the lowest concentration that resulted Synthesis 5 (2007) 773e778.
in 90% of reduction of growth. Itraconazole was used as positive [17] Z. Zhou, Z.-X. Lin, D. Liang, J.-Q. Hu, First synthesis of ring-B C60-substituted
derivatives of N,N-(tetrachlorophthaloyl)dehydroabietylamine, Tetrahedron
control at final concentrations of 0.031e16 mg/mL. A negative 69 (2013) 43e49.
control (inoculum without treatment) was also included. MIC [18] C.-C. Wen, Y.-H. Kuo, J.-T. Jan, P.-H. Liang, S.-Y. Wang, H.-G. Liu, C.-K. Lee, S.-
values were expressed as geometric mean (GM-MIC) of tests per- T. Chang, C.-J. Kuo, S.-S. Lee, C.-C. Hou, P.-W. Hsiao, S.-C. Chien, L.-F. Shyur, N.-
S. Yang, Specific plant terpenoids and lignoids possess potent antiviral ac-
formed in duplicate in three different assays against each fungi tivities against severe acute respiratory syndrome coronavirus, J. Med. Chem.
species. 50 (2007) 4087e4095.
[19] (a) J. Becerra, C. Flores, J. Mena, P. Aqueveque, J. Alarco  n, M. Bittner,
V. Herna ndez, M. Hoeneisen, E. Ruiz, M. Silva, Antifungal and antibacterial
Competing interests activity of diterpenes isolated from wood extractables of Chilean podo-
carpaceae, Bol. Soc. Chil. Quim. 47 (2002) 151e157;
One patent application based on these results has been sub- (b) M. Fronza, R. Murillo, S. Slusarczyk, M. Adams, M. Hamburguer,
B. Heinzmann, S. Laufer, I. Merfort, In vitro cytotoxic activity of abietane
mitted by M.A.G. and L.A.B.-G. as inventors.
diterpenes from peltodon longipes as well as salvia miltiorrhiza and salvia
sahendica, Bioorg. Med. Chem. 19 (2011) 4876e4881.
Acknowledgments [20] M. Bispo de Jesus, W.F. Zambuzzi, R.R. Ruela de Sousa, C. Areche, A.C. Santos
de Souza, H. Aoyama, G. Schmeda-Hirschmann, J.A. Rodriguez, A.R.M. de
Souza Brito, M.P. Peppelenbosch, J. den Hertog, E. de Paula, C.V. Ferreira,
Financial support from COLCIENCIAS-Grant RC-366-2011 and Ferruginol suppresses survival signaling pathways in androgen-independent
111554531592 (Patrimonio Auto  nomo del Fondo Nacional de human prostate cancer cells, Biochimie 90 (2008) 843e854.
88 V.C. Roa-Linares et al. / European Journal of Medicinal Chemistry 108 (2016) 79e88

[21] C. Krummenacher, A. Carfí, R.J. Eisenberg, G.H. Cohen, Entry of herpesviruses (2008) 113e118.
into cells: the enigma variations, Adv. Exp. Med. Biol. 790 (2013) 178e195. [30] M.P. Taylor, O.O. Koyuncu, L.W. Enquist, Subversion of the actin cytoskeleton
[22] D. Chattopadhyay, M.C. Sarkar, T. Chatterjee, R. Sharma Dey, P. Bag, during viral infection, Nat. Rev. Microbiol. 9 (2011) 427e439.
S. Chakraborti, M.T. Khan, Recent advancements for the evaluation of anti- [31] J.-L. Wang, J.-L. Zhang, W. Chen, X.-F. Xu, N. Gao, D.-Y. Fan, J. An, Roles of small
viral activities of natural products, N. Biotechnol. 25 (2009) 347e368. GTPase Rac1 in the regulation of actin cytoskeleton during dengue virus
[23] P. Zhong, L.M. Agosto, J.B. Munro, W. Mothes, Cell-to-cell transmission of vi- infection, PLoS Negl. Trop. Dis. 4 (2010) e809.
ruses, Curr. Opin. Virol. 3 (2013) 44e50. [32] M. Martínez-Gutierrez, J.E. Castellanos, J.C. Gallego-Go mez, Statins reduce
[24] L.T. Lin, T.Y. Chen, C.Y. Chung, R.S. Noyce, T.B. Grindley, C. McCormick, T.C. Lin, dengue virus production via decreased virion assembly, Intervirology 54
G.H. Wang, C.C. Lin, C.D. Richardson, Hydrolyzable Tannins (Chebulagic Acid (2011) 202e216.
and Punicalagin) Target Viral glycoprotein-glycosaminoglycan interactions to , A. Lasure, D. Vanden Berghe, P.C. Rwangabo,
[33] A.J. Vlietinck, L. Van Hoof, J. Totte
inhibit herpes simplex virus 1 entry and cell-to-cell spread, J. Virol. 85 (2011) J. Myukiyumwami, Screening of hundred Rwandese medicinal plants for
4386e4398. antimicrobial and antiviral properties, J. Ethnopharm. 46 (1995) 31e47.
[25] (a) K. Nyberg, M. Ekblad, T. Bergstro €m, C. Freeman, C.R. Parish, V. Ferro, [34] L.I.C. Tang, A.P.K. Ling, R.Y. Koh, S.M. Chye, K.G.L. Voon, Screening of anti-
E. Trybala, The low molecular weight heparan sulfate-mimetic, PI-88, inhibits dengue activity in methanolic extracts of medicinal plants, BMC Comple-
cell-to-cell spread of herpes simplex virus, Antivir. Res. 63 (2004) 15e24; mentary Altern. Med. 12 (3) (2012).
(b) M. Ekblad, B. Adamiak, T. Bergstrom, K.D. Johnstone, T. Karoli, L. Liu, [35] F.T. Cardozo, C.M. Camelini, A. Mascarello, M.J. Rossi, R.J. Nunes, C.R. Barardi,
V. Ferro, E. Trybala, A highly lipophilic sulfated tetrasaccharide glycoside M.M. de Mendonça, C.M. Simo ~es, Antiherpetic activity of a sulfated poly-
related to muparfostat (PI-88) exhibits virucidal activity against herpes sim- saccharide from agaricus brasiliensis mycelia, Antivir. Res. 92 (2011)
plex virus, Antivir. Res. 86 (2010) 196e203. 108e114.
[26] K. Maringer, J. Stylianou, G. Elliott, A network of protein interactions around [36] NCCLS, Reference method for broth dilution antifungal susceptibility testing of
the herpes simplex virus tegument protein VP22, J. Virol. 86 (2012) conidial forming filamentous fungi, Approved standard NCCLS M38-A, Na-
12971e12982. tional committee for clinical Laboratory Standards, Wayne, 2002.
[27] D.C. Johnson, M. Webb, T.W. Wisner, C. Brunetti, Herpes simplex virus gE/gI [37] M. Cuenca-Estrella, C.B. Moore, F. Barchiesi, J. Bille, E. Chryssanthou,
sorts nascent virions to epithelial cell junctions, promoting virus spread, D.W. Denning, J.P. Donnelly, F. Dromer, B. Dupont, J.H. Rex, M.D. Richardson,
J. Virol. 75 (2001) 821e833. B. Sancak, P.E. Verweij, J.L. Rodríguez-Tudela, Multicenter evaluation of the
[28] R.M. Mingo, J. Han, W.W. Newcomb, J.C. Brown, Replication of herpes simplex reproducibility of the proposed antifungal susceptibility testing method for
virus: egress of progeny virus at specialized cell membrane sites, J. Virol. 86 fermentative yeasts of the antifungal susceptibility testing subcommittee of
(2012) 7084e7097. the European committee on antimicrobial susceptibility testing (AFST-
[29] R. Dixit, V. Tiwari, D. Shukla, Herpes simplex virus type 1 induces filopodia in EUCAST), Clin. Microbiol. Infect. 9 (2003) 467e474.
differentiated P19 neural cells to facilitate viral spread, Neurosci. Lett. 440

Вам также может понравиться